1
|
Sorrentino U, O'Neill AG, Kollman JM, Jinnah HA, Zech M. Purine Metabolism and Dystonia: Perspectives of a Long-Promised Relationship. Ann Neurol 2025; 97:809-825. [PMID: 40026236 PMCID: PMC12010064 DOI: 10.1002/ana.27227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 03/05/2025]
Abstract
Dystonia research focuses on the identification of converging biological pathways, allowing to define molecular drivers that serve as treatment targets. We summarize evidence supporting the concept that aberrations in purine metabolism intersect with dystonia pathogenesis. The recent discovery of IMPDH2-related dystonia introduced a gain-of-function paradigm in purinergic system defects, offering new perspectives to understand purine-pool imbalances in brain diseases. We discuss commonalities between known dystonia-linked mechanisms and mechanisms emerging from studies of purine metabolism disorders including Lesch-Nyhan disease. Together, we hypothesize that a greater appreciation of the relevance of purine perturbances in dystonia can offer fresh avenues for therapeutic intervention. ANN NEUROL 2025;97:809-825.
Collapse
Affiliation(s)
- Ugo Sorrentino
- Institute of Human Genetics, Technical University of Munich, School of Medicine and HealthMunichGermany
| | | | | | - Hyder A. Jinnah
- Departments of Neurology, Human Genetics and PediatricsEmory University School of MedicineAtlantaGA
| | - Michael Zech
- Institute of Human Genetics, Technical University of Munich, School of Medicine and HealthMunichGermany
- Institute of Neurogenomics, Helmholtz MunichNeuherbergGermany
- Institute for Advanced Study, Technical University of MunichGarchingGermany
| |
Collapse
|
2
|
Olatoke T, Zhang EY, Wagner A, He Q, Li S, Astreinidis A, McCormack FX, Xu Y, Yu JJ. STAT1 Promotes PD-L1 Activation and Tumor Growth in Lymphangioleiomyomatosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.11.627871. [PMID: 39713456 PMCID: PMC11661278 DOI: 10.1101/2024.12.11.627871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Lymphangioleiomyomatosis (LAM) is a cystic lung disease that primarily affects women. LAM is caused by the invasion of metastatic smooth muscle-like cells into the lung parenchyma, leading to abnormal cell proliferation, lung remodeling and progressive respiratory failure. LAM cells have TSC gene mutations, which occur sporadically or in people with Tuberous Sclerosis Complex. Although it is known that hyperactivation of the mechanistic target of rapamycin complex 1 (mTORC1) due to TSC2 gene mutations contributes to aberrant cell growth in LAM lung, tumor origin and invasive mechanism remain unclear. To determine molecular drivers responsible for aberrant LAM cell growth, we performed integrative single-cell transcriptomic analysis and predicted that STAT1 interacts with Pre-B cell leukemia transcription factor (PBX1) to regulate LAM cell survival. Here, we show activation of STAT1 and STAT3 proteins in TSC2-deficient LAM models. Fludarabine, a potent STAT1 inhibitor, induced the death of TSC2-deficient cells, increased caspase-3 cleavage, and phosphorylation of necroptosis marker RIP1. Fludarabine treatment impeded lung colonization of TSC2-deficient cells and uterine tumor progression, associated with reduced percentage of PCNA-positive cells in vivo. Interestingly, IFN-γ treatment increased STAT1 phosphorylation and PD-L1 expression, indicating that STAT1 aids TSC2-deficient tumor cells in evading immune surveillance in LAM. Our findings indicate that STAT1 signaling is critical for LAM cell survival and could be targeted to treat LAM and other mTORC1 hyperactive tumors.
Collapse
Affiliation(s)
- Tasnim Olatoke
- Department of Pharmacology, Physiology and Neurobiology, University of Cincinnati; Cincinnati, OH 45267, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of Cincinnati; Cincinnati, OH 45267, USA
| | - Erik Y Zhang
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of Cincinnati; Cincinnati, OH 45267, USA
| | - Andrew Wagner
- Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229, USA
| | - Quan He
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of Cincinnati; Cincinnati, OH 45267, USA
| | - Siru Li
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of Cincinnati; Cincinnati, OH 45267, USA
| | - Aristotelis Astreinidis
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of Cincinnati; Cincinnati, OH 45267, USA
| | - Francis X McCormack
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of Cincinnati; Cincinnati, OH 45267, USA
| | - Yan Xu
- Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center; Cincinnati, OH 45229, USA
| | - Jane J Yu
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of Cincinnati; Cincinnati, OH 45267, USA
| |
Collapse
|
3
|
Wu D, Yang S, Yuan C, Zhang K, Tan J, Guan K, Zeng H, Huang C. Targeting purine metabolism-related enzymes for therapeutic intervention: A review from molecular mechanism to therapeutic breakthrough. Int J Biol Macromol 2024; 282:136828. [PMID: 39447802 DOI: 10.1016/j.ijbiomac.2024.136828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 10/02/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
Purines are ancient metabolites with established and emerging metabolic and non-metabolic signaling attributes. The expression of purine metabolism-related genes is frequently activated in human malignancies, correlating with increased cancer aggressiveness and chemoresistance. Importantly, under certain stimulating conditions, the purine biosynthetic enzymes can assemble into a metabolon called "purinosomes" to enhance purine flux. Current evidence suggests that purine flux is regulated by a complex circuit that encompasses transcriptional, post-translational, metabolic, and association-dependent regulatory mechanisms. Furthermore, purines within the tumor microenvironment modulate cancer immunity through signaling mediated by purinergic receptors. The deregulation of purine metabolism has significant metabolic consequences, particularly hyperuricemia. Herbal-based therapeutics have emerged as valuable pharmacological interventions for the treatment of hyperuricemia by inhibiting the activity of hepatic XOD, modulating the expression of renal urate transporters, and suppressing inflammatory responses. This review summarizes recent advancements in the understanding of purine metabolism in clinically relevant malignancies and metabolic disorders. Additionally, we discuss the role of herbal interventions and the interaction between the host and gut microbiota in the regulation of purine homeostasis. This information will fuel the innovation of therapeutic strategies that target the disease-associated rewiring of purine metabolism for therapeutic applications.
Collapse
Affiliation(s)
- Di Wu
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Shengqiang Yang
- School of Basic Medicine, Youjiang Medical University for Nationalities, Baise 533000, China
| | - Chenyang Yuan
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Kejia Zhang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Jiachen Tan
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Kaifeng Guan
- School of Advanced Agricultural Sciences, Peking University, Beijing 100871, China.
| | - Hong Zeng
- School of Basic Medicine, Youjiang Medical University for Nationalities, Baise 533000, China.
| | - Chunjie Huang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China.
| |
Collapse
|
4
|
Nengroo MA, Ben-Sahra I. Brain-penetrating molecule might offer a route to treat glioblastoma tumours. Nature 2024; 636:307-308. [PMID: 39567800 DOI: 10.1038/d41586-024-03556-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
|
5
|
Chen YJ, Iyer SV, Hsieh DCC, Li B, Elias HK, Wang T, Li J, Ganbold M, Lien MC, Peng YC, Xie XP, Jayewickreme CD, van den Brink MRM, Brady SF, Lim SK, Parada LF. Gliocidin is a nicotinamide-mimetic prodrug that targets glioblastoma. Nature 2024; 636:466-473. [PMID: 39567689 PMCID: PMC11665509 DOI: 10.1038/s41586-024-08224-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/15/2024] [Indexed: 11/22/2024]
Abstract
Glioblastoma is incurable and in urgent need of improved therapeutics1. Here we identify a small compound, gliocidin, that kills glioblastoma cells while sparing non-tumour replicative cells. Gliocidin activity targets a de novo purine synthesis vulnerability in glioblastoma through indirect inhibition of inosine monophosphate dehydrogenase 2 (IMPDH2). IMPDH2 blockade reduces intracellular guanine nucleotide levels, causing nucleotide imbalance, replication stress and tumour cell death2. Gliocidin is a prodrug that is anabolized into its tumoricidal metabolite, gliocidin-adenine dinucleotide (GAD), by the enzyme nicotinamide nucleotide adenylyltransferase 1 (NMNAT1) of the NAD+ salvage pathway. The cryo-electron microscopy structure of GAD together with IMPDH2 demonstrates its entry, deformation and blockade of the NAD+ pocket3. In vivo, gliocidin penetrates the blood-brain barrier and extends the survival of mice with orthotopic glioblastoma. The DNA alkylating agent temozolomide induces Nmnat1 expression, causing synergistic tumour cell killing and additional survival benefit in orthotopic patient-derived xenograft models. This study brings gliocidin to light as a prodrug with the potential to improve the survival of patients with glioblastoma.
Collapse
Affiliation(s)
- Yu-Jung Chen
- Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Swathi V Iyer
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David Chun-Cheng Hsieh
- Tri-Institutional PhD Program in Chemical Biology, The Rockefeller University, New York, NY, USA
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, New York, NY, USA
| | - Buren Li
- Structure Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Harold K Elias
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- National Institutes of Health, Bethesda, MD, USA
| | - Tao Wang
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Neuroscience Program, Weill Cornell Medicine Graduate School of Medical Sciences, New York, NY, USA
| | - Jing Li
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mungunsarnai Ganbold
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michelle C Lien
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yu-Chun Peng
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Xuanhua P Xie
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chenura D Jayewickreme
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Sean F Brady
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, New York, NY, USA
| | - S Kyun Lim
- KOBIOLABS, Inc., Seongnam-si, South Korea
| | - Luis F Parada
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
6
|
Manning BD, Dibble CC. Growth Signaling Networks Orchestrate Cancer Metabolic Networks. Cold Spring Harb Perspect Med 2024; 14:a041543. [PMID: 38438221 PMCID: PMC11444256 DOI: 10.1101/cshperspect.a041543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Normal cells grow and divide only when instructed to by signaling pathways stimulated by exogenous growth factors. A nearly ubiquitous feature of cancer cells is their capacity to grow independent of such signals, in an uncontrolled, cell-intrinsic manner. This property arises due to the frequent oncogenic activation of core growth factor signaling pathway components, including receptor tyrosine kinases, PI3K-AKT, RAS-RAF, mTORC1, and MYC, leading to the aberrant propagation of pro-growth signals independent of exogenous growth factors. The growth of both normal and cancer cells requires the acquisition of nutrients and their anabolic conversion to the primary macromolecules underlying biomass production (protein, nucleic acids, and lipids). The core growth factor signaling pathways exert tight regulation of these metabolic processes and the oncogenic activation of these pathways drive the key metabolic properties of cancer cells and tumors. Here, we review the molecular mechanisms through which these growth signaling pathways control and coordinate cancer metabolism.
Collapse
Affiliation(s)
- Brendan D Manning
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Christian C Dibble
- Department of Pathology, Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
7
|
Allegrini S, Camici M, Garcia-Gil M, Pesi R, Tozzi MG. Interplay between mTOR and Purine Metabolism Enzymes and Its Relevant Role in Cancer. Int J Mol Sci 2024; 25:6735. [PMID: 38928439 PMCID: PMC11203890 DOI: 10.3390/ijms25126735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/14/2024] [Accepted: 06/16/2024] [Indexed: 06/28/2024] Open
Abstract
Tumor cells reprogram their metabolism to meet the increased demand for nucleotides and other molecules necessary for growth and proliferation. In fact, cancer cells are characterized by an increased "de novo" synthesis of purine nucleotides. Therefore, it is not surprising that specific enzymes of purine metabolism are the targets of drugs as antineoplastic agents, and a better knowledge of the mechanisms underlying their regulation would be of great help in finding new therapeutic approaches. The mammalian target of the rapamycin (mTOR) signaling pathway, which is often activated in cancer cells, promotes anabolic processes and is a major regulator of cell growth and division. Among the numerous effects exerted by mTOR, noteworthy is its empowerment of the "de novo" synthesis of nucleotides, accomplished by supporting the formation of purinosomes, and by increasing the availability of necessary precursors, such as one-carbon formyl group, bicarbonate and 5-phosphoribosyl-1-pyrophosphate. In this review, we highlight the connection between purine and mitochondrial metabolism, and the bidirectional relation between mTOR signaling and purine synthesis pathways.
Collapse
Affiliation(s)
- Simone Allegrini
- Unità di Biochimica, Dipartimento di Biologia, Università di Pisa, Via San Zeno 51, 56127 Pisa, Italy; (M.C.); (R.P.); (M.G.T.)
- Centro di Ricerca Interdipartimentale Nutrafood “Nutraceuticals and Food for Health”, Università di Pisa, 56126 Pisa, Italy;
- CISUP, Centro per l’Integrazione Della Strumentazione Dell’Università di Pisa, 56127 Pisa, Italy
| | - Marcella Camici
- Unità di Biochimica, Dipartimento di Biologia, Università di Pisa, Via San Zeno 51, 56127 Pisa, Italy; (M.C.); (R.P.); (M.G.T.)
| | - Mercedes Garcia-Gil
- Centro di Ricerca Interdipartimentale Nutrafood “Nutraceuticals and Food for Health”, Università di Pisa, 56126 Pisa, Italy;
- CISUP, Centro per l’Integrazione Della Strumentazione Dell’Università di Pisa, 56127 Pisa, Italy
- Unità di Fisiologia Generale, Dipartimento di Biologia, Università di Pisa, Via San Zeno 31, 56127 Pisa, Italy
| | - Rossana Pesi
- Unità di Biochimica, Dipartimento di Biologia, Università di Pisa, Via San Zeno 51, 56127 Pisa, Italy; (M.C.); (R.P.); (M.G.T.)
| | - Maria Grazia Tozzi
- Unità di Biochimica, Dipartimento di Biologia, Università di Pisa, Via San Zeno 51, 56127 Pisa, Italy; (M.C.); (R.P.); (M.G.T.)
| |
Collapse
|
8
|
Tangudu NK, Buj R, Wang H, Wang J, Cole AR, Uboveja A, Fang R, Amalric A, Yang B, Chatoff A, Crispim CV, Sajjakulnukit P, Lyons MA, Cooper K, Hempel N, Lyssiotis CA, Chandran UR, Snyder NW, Aird KM. De Novo Purine Metabolism is a Metabolic Vulnerability of Cancers with Low p16 Expression. CANCER RESEARCH COMMUNICATIONS 2024; 4:1174-1188. [PMID: 38626341 PMCID: PMC11064835 DOI: 10.1158/2767-9764.crc-23-0450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/04/2024] [Accepted: 04/11/2024] [Indexed: 04/18/2024]
Abstract
p16 is a tumor suppressor encoded by the CDKN2A gene whose expression is lost in approximately 50% of all human cancers. In its canonical role, p16 inhibits the G1-S-phase cell cycle progression through suppression of cyclin-dependent kinases. Interestingly, p16 also has roles in metabolic reprogramming, and we previously published that loss of p16 promotes nucleotide synthesis via the pentose phosphate pathway. However, the broader impact of p16/CDKN2A loss on other nucleotide metabolic pathways and potential therapeutic targets remains unexplored. Using CRISPR knockout libraries in isogenic human and mouse melanoma cell lines, we determined several nucleotide metabolism genes essential for the survival of cells with loss of p16/CDKN2A. Consistently, many of these genes are upregulated in melanoma cells with p16 knockdown or endogenously low CDKN2A expression. We determined that cells with low p16/CDKN2A expression are sensitive to multiple inhibitors of de novo purine synthesis, including antifolates. Finally, tumors with p16 knockdown were more sensitive to the antifolate methotrexate in vivo than control tumors. Together, our data provide evidence to reevaluate the utility of these drugs in patients with p16/CDKN2Alow tumors as loss of p16/CDKN2A may provide a therapeutic window for these agents. SIGNIFICANCE Antimetabolites were the first chemotherapies, yet many have failed in the clinic due to toxicity and poor patient selection. Our data suggest that p16 loss provides a therapeutic window to kill cancer cells with widely-used antifolates with relatively little toxicity.
Collapse
Affiliation(s)
- Naveen Kumar Tangudu
- Department of Pharmacology and Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Raquel Buj
- Department of Pharmacology and Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Hui Wang
- Department of Pharmacology and Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jiefei Wang
- Department of Biomedical Informatics and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Aidan R. Cole
- Department of Pharmacology and Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Apoorva Uboveja
- Department of Pharmacology and Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Richard Fang
- Department of Pharmacology and Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Amandine Amalric
- Department of Pharmacology and Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Baixue Yang
- Department of Pharmacology and Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Tsinghua University School of Medicine, Beijing, P.R. China
| | - Adam Chatoff
- Department of Cardiovascular Sciences, Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Claudia V. Crispim
- Department of Cardiovascular Sciences, Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Peter Sajjakulnukit
- Department of Molecular and Integrative Physiology, Department of Internal Medicine, Division of Gastroenterology, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Maureen A. Lyons
- Genomics Facility, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Kristine Cooper
- Biostatistics Facility, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Nadine Hempel
- Division of Hematology/Oncology, Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Costas A. Lyssiotis
- Department of Molecular and Integrative Physiology, Department of Internal Medicine, Division of Gastroenterology, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Uma R. Chandran
- Department of Biomedical Informatics and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Nathaniel W. Snyder
- Department of Cardiovascular Sciences, Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Katherine M. Aird
- Department of Pharmacology and Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
9
|
Paul R, Shreya S, Pandey S, Shriya S, Abou Hammoud A, Grosset CF, Prakash Jain B. Functions and Therapeutic Use of Heat Shock Proteins in Hepatocellular Carcinoma. LIVERS 2024; 4:142-163. [DOI: 10.3390/livers4010011] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/10/2024] Open
Abstract
Heat shock proteins are intracellular proteins expressed in prokaryotes and eukaryotes that help protect the cell from stress. They play an important role in regulating cell cycle and cell death, work as molecular chaperons during the folding of newly synthesized proteins, and also in the degradation of misfolded proteins. They are not only produced under stress conditions like acidosis, energy depletion, and oxidative stress but are also continuously synthesized as a result of their housekeeping functions. There are different heat shock protein families based on their molecular weight, like HSP70, HSP90, HSP60, HSP27, HSP40, etc. Heat shock proteins are involved in many cancers, particularly hepatocellular carcinoma, the main primary tumor of the liver in adults. Their deregulations in hepatocellular carcinoma are associated with metastasis, angiogenesis, cell invasion, and cell proliferation and upregulated heat shock proteins can be used as either diagnostic or prognostic markers. Targeting heat shock proteins is a relevant strategy for the treatment of patients with liver cancer. In this review, we provide insights into heat shock proteins and heat shock protein-like proteins (clusterin) in the progression of hepatocellular carcinoma and their use as therapeutic targets.
Collapse
Affiliation(s)
- Ramakrushna Paul
- Gene Expression and Signaling Lab, Department of Zoology, Mahatma Gandhi Central University, Motihari 845401, India
| | - Smriti Shreya
- Gene Expression and Signaling Lab, Department of Zoology, Mahatma Gandhi Central University, Motihari 845401, India
| | | | - Srishti Shriya
- Gene Expression and Signaling Lab, Department of Zoology, Mahatma Gandhi Central University, Motihari 845401, India
| | - Aya Abou Hammoud
- MIRCADE Team, U1312, Bordeaux Institute of Oncology, BRIC, INSERM, University of Bordeaux, 33000 Bordeaux, France
| | - Christophe F. Grosset
- MIRCADE Team, U1312, Bordeaux Institute of Oncology, BRIC, INSERM, University of Bordeaux, 33000 Bordeaux, France
| | - Buddhi Prakash Jain
- Gene Expression and Signaling Lab, Department of Zoology, Mahatma Gandhi Central University, Motihari 845401, India
| |
Collapse
|
10
|
Xie Q, Cao Z, You W, Cai X, Shen M, Yin Z, Jiang Y, Wang X, Ye S. Ganodermanontriol Suppresses the Progression of Lung Adenocarcinoma by Activating CES2 to Enhance the Metabolism of Mycophenolate Mofetil. J Microbiol Biotechnol 2024; 34:249-261. [PMID: 38419324 PMCID: PMC10940751 DOI: 10.4014/jmb.2306.06020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 03/02/2024]
Abstract
New anti-lung cancer therapies are urgently required to improve clinical outcomes. Since ganodermanontriol (GDNT) has been identified as a potential antineoplastic agent, its role in lung adenocarcinoma (LUAD) is investigated in this study. Concretely, lung cancer cells were treated with GDNT and/or mycophenolate mofetil (MMF), after which MTT assay, flow cytometry and Western blot were conducted. Following bioinformatics analysis, carboxylesterase 2 (CES2) was knocked down and rescue assays were carried out in vitro. Xenograft experiment was performed on mice, followed by drug administration, measurement of tumor growth and determination of CES2, IMPDH1 and IMPDH2 expressions. As a result, the viability of lung cancer cells was reduced by GDNT or MMF. GDNT enhanced the effects of MMF on suppressing viability, promoting apoptosis and inducing cell cycle arrest in lung cancer cells. GDNT up-regulated CES2 level, and strengthened the effects of MMF on down-regulating IMPDH1 and IMPDH2 levels in the cells. IMPDH1 and IMPDH2 were highly expressed in LUAD samples. CES2 was a potential target for GDNT. CES2 knockdown reversed the synergistic effect of GDNT and MMF against lung cancer in vitro. GDNT potentiated the role of MMF in inhibiting tumor growth and expressions of CES2 and IMPDH1/2 in lung cancer in vivo. Collectively, GDNT suppresses the progression of LUAD by activating CES2 to enhance the metabolism of MMF.
Collapse
Affiliation(s)
- Qingfeng Xie
- Respiratory Department, Longquan People’s Hospital, No. 699, Dongcha Road, Longquan City, Zhejiang Province, 323000, P.R. China
| | - Zhuo Cao
- Respiratory Department, The Sixth Affiliated Hospital of Wenzhou Medical University, No. 15 Dazhong Street, Liandu District, Lishui City, Zhejiang Province, 323000, P.R. China
| | - Weiling You
- Respiratory Department, Longquan People’s Hospital, No. 699, Dongcha Road, Longquan City, Zhejiang Province, 323000, P.R. China
| | - Xiaoping Cai
- Respiratory Department, The Sixth Affiliated Hospital of Wenzhou Medical University, No. 15 Dazhong Street, Liandu District, Lishui City, Zhejiang Province, 323000, P.R. China
| | - Mei Shen
- Longquan People’s Hospital, No. 699, Dongcha Road, Longquan City, Zhejiang Province, 323000, P.R. China
| | - Zhangyong Yin
- Respiratory Department, The Sixth Affiliated Hospital of Wenzhou Medical University, No. 15 Dazhong Street, Liandu District, Lishui City, Zhejiang Province, 323000, P.R. China
| | - Yiwei Jiang
- Wenzhou Medical University, Wenzhou Chashan Higher Education Park, Wenzhou, Zhejiang Province, 325006, P.R. China
| | - Xin Wang
- Wenzhou Medical University, Wenzhou Chashan Higher Education Park, Wenzhou, Zhejiang Province, 325006, P.R. China
| | - Siyu Ye
- School of Public Administration, Wenzhou Medical University, Wenzhou Chashan Higher Education Park, Wenzhou, Zhejiang Province, 325006, P.R. China
| |
Collapse
|
11
|
Zhou W, Zhao Z, Lin A, Yang JZ, Xu J, Wilder-Romans K, Yang A, Li J, Solanki S, Speth JM, Walker N, Scott AJ, Wang L, Wen B, Andren A, Zhang L, Kothari AU, Yao Y, Peterson ER, Korimerla N, Werner CK, Ullrich A, Liang J, Jacobson J, Palavalasa S, O’Brien AM, Elaimy AL, Ferris SP, Zhao SG, Sarkaria JN, Győrffy B, Zhang S, Al-Holou WN, Umemura Y, Morgan MA, Lawrence TS, Lyssiotis CA, Peters-Golden M, Shah YM, Wahl DR. GTP Signaling Links Metabolism, DNA Repair, and Responses to Genotoxic Stress. Cancer Discov 2024; 14:158-175. [PMID: 37902550 PMCID: PMC10872631 DOI: 10.1158/2159-8290.cd-23-0437] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 09/07/2023] [Accepted: 10/19/2023] [Indexed: 10/31/2023]
Abstract
How cell metabolism regulates DNA repair is incompletely understood. Here, we define a GTP-mediated signaling cascade that links metabolism to DNA repair and has significant therapeutic implications. GTP, but not other nucleotides, regulates the activity of Rac1, a guanine nucleotide-binding protein, which promotes the dephosphorylation of serine 323 on Abl-interactor 1 (Abi-1) by protein phosphatase 5 (PP5). Dephosphorylated Abi-1, a protein previously not known to activate DNA repair, promotes nonhomologous end joining. In patients and mouse models of glioblastoma, Rac1 and dephosphorylated Abi-1 mediate DNA repair and resistance to standard-of-care genotoxic treatments. The GTP-Rac1-PP5-Abi-1 signaling axis is not limited to brain cancer, as GTP supplementation promotes DNA repair and Abi-1-S323 dephosphorylation in nonmalignant cells and protects mouse tissues from genotoxic insult. This unexpected ability of GTP to regulate DNA repair independently of deoxynucleotide pools has important implications for normal physiology and cancer treatment. SIGNIFICANCE A newly described GTP-dependent signaling axis is an unexpected link between nucleotide metabolism and DNA repair. Disrupting this pathway can overcome cancer resistance to genotoxic therapy while augmenting it can mitigate genotoxic injury of normal tissues. This article is featured in Selected Articles from This Issue, p. 5.
Collapse
Affiliation(s)
- Weihua Zhou
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Zitong Zhao
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Department of Oncology, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shan Xi, PR China
| | - Angelica Lin
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - John Z Yang
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Jie Xu
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Kari Wilder-Romans
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Annabel Yang
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Jing Li
- Cell Signaling Technology, Inc., Danvers, MA, USA
| | - Sumeet Solanki
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Jennifer M Speth
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Natalie Walker
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Andrew J Scott
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Lu Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
| | - Bo Wen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
| | - Anthony Andren
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Li Zhang
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Ayesha U Kothari
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Yangyang Yao
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Department of Oncology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, PR China
| | - Erik R Peterson
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Navyateja Korimerla
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Christian K Werner
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Alexander Ullrich
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Jessica Liang
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Janna Jacobson
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Sravya Palavalasa
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Alexandra M O’Brien
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Ameer L Elaimy
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Sean P Ferris
- Department of Pathology, Division of Neuropathology, University of Michigan, Ann Arbor, MI, USA
| | - Shuang G Zhao
- Department of Human Oncology, University of Wisconsin Madison, WI, USA
| | | | - Balázs Győrffy
- Department of Bioinformatics, Semmelweis University, Budapest, Hungary; and TTK Cancer Biomarker Research Group, Institute of Enzymology, Budapest, Hungary
| | - Shuqun Zhang
- Department of Oncology, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shan Xi, PR China
| | - Wajd N Al-Holou
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, United States
| | - Yoshie Umemura
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Meredith A Morgan
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Theodore S Lawrence
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Costas A Lyssiotis
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology and Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Marc Peters-Golden
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Yatrik M Shah
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Daniel R Wahl
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
- Lead contact
| |
Collapse
|
12
|
Hassan AMS, Elfiky AA, Elgohary AM. Triple in silico targeting of IMPDH enzyme and RNA-dependent RNA polymerase of both SARS-CoV-2 and Rhizopus oryzae. Future Microbiol 2024; 19:9-19. [PMID: 38294272 DOI: 10.2217/fmb-2023-0103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 08/23/2023] [Indexed: 02/01/2024] Open
Abstract
Aim: Mucormycosis has been associated with SARS-CoV-2 infections during the last year. The aim of this study was to triple-hit viral and fungal RNA-dependent RNA polymerases (RdRps) and human inosine monophosphate dehydrogenase (IMPDH). Materials & methods: Molecular docking and molecular dynamics simulation were used to test nucleotide inhibitors (NIs) against the RdRps of SARS-CoV-2 and Rhizopus oryzae RdRp. These same inhibitors targeted IMPDH. Results: Four NIs revealed a comparable binding affinity to the two drugs, remdesivir and sofosbuvir. Binding energies were calculated using the most abundant conformations of the RdRps after 100-ns molecular dynamics simulation. Conclusion: We suggest the triple-inhibition potential of four NIs against pathogenic RdRps and IMPDH, which is worth experimental validation.
Collapse
Affiliation(s)
| | - Abdo A Elfiky
- Biophysics Department, Faculty of Sciences, Cairo University, Giza, Dokki, 12613, Egypt
| | - Alaa M Elgohary
- Biophysics Department, Faculty of Sciences, Cairo University, Giza, Dokki, 12613, Egypt
| |
Collapse
|
13
|
Walczak J, Iwaszkiewicz-Grześ D, Cholewiński G. Approaches Towards Better Immunosuppressive Agents. Curr Top Med Chem 2024; 24:1230-1263. [PMID: 38561615 DOI: 10.2174/0115680266292661240322072908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 03/12/2024] [Accepted: 03/12/2024] [Indexed: 04/04/2024]
Abstract
Several classes of compounds are applied in clinics due to their immunosuppressive properties in transplantology and the treatment of autoimmune diseases. Derivatives of mycophenolic acid, corticosteroids and chemotherapeutics bearing heterocyclic moieties like methotrexate, azathioprine, mizoribine, and ruxolitinib are active substances with investigated mechanisms of action. However, improved synthetic approaches of known drugs and novel derivatives are still being reported to attempt better accessibility and therapeutic properties. In this review article, we present the synthesis of the designed chemical structures based on recent literature reports concerning novel compounds as promising immunosuppressive drugs. Moreover, some of the discussed derivers revealed also other types of activities with prospective medicinal potential.
Collapse
Affiliation(s)
- Juliusz Walczak
- Department of Organic Chemistry, Faculty of Chemistry, Gdańsk University of Technology, G. Narutowicza 11/12, 80-233, Gdańsk, Poland
| | - Dorota Iwaszkiewicz-Grześ
- Department of Medical Immunology, Faculty of Medicine, Medical University of Gdansk, ul. Dębinki 7, 80-210, Gdańsk, Poland
| | - Grzegorz Cholewiński
- Department of Organic Chemistry, Faculty of Chemistry, Gdańsk University of Technology, G. Narutowicza 11/12, 80-233, Gdańsk, Poland
| |
Collapse
|
14
|
Tangudu NK, Buj R, Wang H, Wang J, Cole AR, Uboveja A, Fang R, Amalric A, Sajjakulnukit P, Lyons MA, Cooper K, Hempel N, Snyder NW, Lyssiotis CA, Chandran UR, Aird KM. De novo purine metabolism is a metabolic vulnerability of cancers with low p16 expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.15.549149. [PMID: 37503050 PMCID: PMC10369956 DOI: 10.1101/2023.07.15.549149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
p16 is a tumor suppressor encoded by the CDKN2A gene whose expression is lost in ~50% of all human cancers. In its canonical role, p16 inhibits the G1-S phase cell cycle progression through suppression of cyclin dependent kinases. Interestingly, p16 also has roles in metabolic reprogramming, and we previously published that loss of p16 promotes nucleotide synthesis via the pentose phosphate pathway. Whether other nucleotide metabolic genes and pathways are affected by p16/CDKN2A loss and if these can be specifically targeted in p16/CDKN2A-low tumors has not been previously explored. Using CRISPR KO libraries in multiple isogenic human and mouse melanoma cell lines, we determined that many nucleotide metabolism genes are negatively enriched in p16/CDKN2A knockdown cells compared to controls. Indeed, many of the genes that are required for survival in the context of low p16/CDKN2A expression based on our CRISPR screens are upregulated in p16 knockdown melanoma cells and those with endogenously low CDKN2A expression. We determined that cells with low p16/Cdkn2a expression are sensitive to multiple inhibitors of de novo purine synthesis, including anti-folates. Tumors with p16 knockdown were more sensitive to the anti-folate methotrexate in vivo than control tumors. Together, our data provide evidence to reevaluate the utility of these drugs in patients with p16/CDKN2A-low tumors as loss of p16/CDKN2A may provide a therapeutic window for these agents.
Collapse
Affiliation(s)
- Naveen Kumar Tangudu
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Raquel Buj
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Hui Wang
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Jiefei Wang
- Department of Biomedical Informatics and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Aidan R. Cole
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Apoorva Uboveja
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Richard Fang
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Amandine Amalric
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Peter Sajjakulnukit
- Department of Molecular and Integrative Physiology, Department of Internal Medicine, Division of Gastroenterology, and Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Maureen A. Lyons
- Genomics Facility UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Kristine Cooper
- Biostatistics Facility UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Nadine Hempel
- Division of Hematology/Oncology, Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA
| | - Nathaniel W. Snyder
- Department of Cardiovascular Sciences, Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Costas A. Lyssiotis
- Department of Molecular and Integrative Physiology, Department of Internal Medicine, Division of Gastroenterology, and Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Uma R. Chandran
- Department of Biomedical Informatics and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Katherine M. Aird
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
15
|
Zhou W, Zhao Z, Lin A, Yang J, Xu J, Kari WR, Yang A, Li J, Solanki S, Speth J, Walker N, Scott AJ, Kothari AU, Yao Y, Peterson ER, Korimerla N, Werner CK, Liang J, Jacobson J, Palavalasa S, Obrien AM, Elaimy AL, Ferris SP, Zhao SG, Sarkaria JN, Győrffy B, Zhang S, Al-Holou WN, Umemura Y, Morgan MA, Lawrence TS, Lyssiotis CA, Peters-Golden M, Shah YM, Wahl DR. GTP signaling links metabolism, DNA repair, and responses to genotoxic stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.12.536297. [PMID: 37090571 PMCID: PMC10120670 DOI: 10.1101/2023.04.12.536297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
How cell metabolism regulates DNA repair is incompletely understood. Here, we define a GTP-mediated signaling cascade that links metabolism to DNA repair and has significant therapeutic implications. GTP, but not other nucleotides, regulates the activity of Rac1, a G protein, that promotes the dephosphorylation of serine 323 on Abl-interactor 1 (Abi-1) by protein phosphatase 5 (PP5). Dephosphorylated Abi-1, a protein previously not known to activate DNA repair, promotes non-homologous end joining. In patients and mouse models of glioblastoma, Rac1 and dephosphorylated Abi-1 mediate DNA repair and resistance to standard of care genotoxic treatments. The GTP-Rac1-PP5-Abi-1 signaling axis is not limited to brain cancer, as GTP supplementation promotes DNA repair and Abi-1-S323 dephosphorylation in non-malignant cells and protects mouse tissues from genotoxic insult. This unexpected ability of GTP to regulate DNA repair independently of deoxynucleotide pools has important implications for normal physiology and cancer treatment.
Collapse
|
16
|
Strzelec M, Detka J, Mieszczak P, Sobocińska MK, Majka M. Immunomodulation—a general review of the current state-of-the-art and new therapeutic strategies for targeting the immune system. Front Immunol 2023; 14:1127704. [PMID: 36969193 PMCID: PMC10033545 DOI: 10.3389/fimmu.2023.1127704] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/23/2023] [Indexed: 03/11/2023] Open
Abstract
In recent years, there has been a tremendous development of biotechnological, pharmacological, and medical techniques which can be implemented in the functional modulation of the immune system components. Immunomodulation has attracted much attention because it offers direct applications in both basic research and clinical therapy. Modulation of a non-adequate, amplified immune response enables to attenuate the clinical course of a disease and restore homeostasis. The potential targets to modulate immunity are as multiple as the components of the immune system, thus creating various possibilities for intervention. However, immunomodulation faces new challenges to design safer and more efficacious therapeutic compounds. This review offers a cross-sectional picture of the currently used and newest pharmacological interventions, genomic editing, and tools for regenerative medicine involving immunomodulation. We reviewed currently available experimental and clinical evidence to prove the efficiency, safety, and feasibility of immunomodulation in vitro and in vivo. We also reviewed the advantages and limitations of the described techniques. Despite its limitations, immunomodulation is considered as therapy itself or as an adjunct with promising results and developing potential.
Collapse
|
17
|
Reyes A, Mohanty A, Pharaon R, Massarelli E. Association between Immunosuppressive Therapy Utilized in the Treatment of Autoimmune Disease or Transplant and Cancer Progression. Biomedicines 2022; 11:biomedicines11010099. [PMID: 36672607 PMCID: PMC9856025 DOI: 10.3390/biomedicines11010099] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 12/28/2022] [Accepted: 12/28/2022] [Indexed: 01/01/2023] Open
Abstract
Autoimmunity and cancer rates have both been on the rise in Western civilization prompting many to investigate the link between the two entities. This review will investigate the complex interactions between the activation and deactivation of the immune system and the development of malignancy. Additional focus will be placed on the main classes of immune inhibitor therapy utilized in transplant patients and in autoimmune disease including TNF-alpha, Calcineurin, mTOR, purine synthesis antagonists and IMPDH inhibitors.
Collapse
|
18
|
McNamara MC, Hosios AM, Torrence ME, Zhao T, Fraser C, Wilkinson M, Kwiatkowski DJ, Henske EP, Wu CL, Sarosiek KA, Valvezan AJ, Manning BD. Reciprocal effects of mTOR inhibitors on pro-survival proteins dictate therapeutic responses in tuberous sclerosis complex. iScience 2022; 25:105458. [PMID: 36388985 PMCID: PMC9663903 DOI: 10.1016/j.isci.2022.105458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 09/30/2022] [Accepted: 10/23/2022] [Indexed: 11/13/2022] Open
Abstract
mTORC1 is aberrantly activated in cancer and in the genetic tumor syndrome tuberous sclerosis complex (TSC), which is caused by loss-of-function mutations in the TSC complex, a negative regulator of mTORC1. Clinically approved mTORC1 inhibitors, such as rapamycin, elicit a cytostatic effect that fails to eliminate tumors and is rapidly reversible. We sought to determine the effects of mTORC1 on the core regulators of intrinsic apoptosis. In TSC2-deficient cells and tumors, we find that mTORC1 inhibitors shift cellular dependence from MCL-1 to BCL-2 and BCL-XL for survival, thereby altering susceptibility to BH3 mimetics that target specific pro-survival BCL-2 proteins. The BCL-2/BCL-XL inhibitor ABT-263 synergizes with rapamycin to induce apoptosis in TSC-deficient cells and in a mouse tumor model of TSC, resulting in a more complete and durable response. These data expose a therapeutic vulnerability in regulation of the apoptotic machinery downstream of mTORC1 that promotes a cytotoxic response to rapamycin.
Collapse
Affiliation(s)
- Molly C. McNamara
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Aaron M. Hosios
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Margaret E. Torrence
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, USA
| | - Ting Zhao
- Department of Urology, Massachusetts General Hospital, Boston, MA, USA
- Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Cameron Fraser
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA 02215, USA
| | - Meghan Wilkinson
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - David J. Kwiatkowski
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Elizabeth P. Henske
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Chin-Lee Wu
- Department of Urology, Massachusetts General Hospital, Boston, MA, USA
- Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Kristopher A. Sarosiek
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA 02215, USA
| | - Alexander J. Valvezan
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, USA
| | - Brendan D. Manning
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
19
|
Zhu J, Wang H, Jiang X. mTORC1 beyond anabolic metabolism: Regulation of cell death. J Biophys Biochem Cytol 2022; 221:213609. [PMID: 36282248 PMCID: PMC9606688 DOI: 10.1083/jcb.202208103] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/03/2022] [Accepted: 10/04/2022] [Indexed: 12/13/2022] Open
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1), a multi-subunit protein kinase complex, interrogates growth factor signaling with cellular nutrient and energy status to control metabolic homeostasis. Activation of mTORC1 promotes biosynthesis of macromolecules, including proteins, lipids, and nucleic acids, and simultaneously suppresses catabolic processes such as lysosomal degradation of self-constituents and extracellular components. Metabolic regulation has emerged as a critical determinant of various cellular death programs, including apoptosis, pyroptosis, and ferroptosis. In this article, we review the expanding knowledge on how mTORC1 coordinates metabolic pathways to impinge on cell death regulation. We focus on the current understanding on how nutrient status and cellular signaling pathways connect mTORC1 activity with ferroptosis, an iron-dependent cell death program that has been implicated in a plethora of human diseases. In-depth understanding of the principles governing the interaction between mTORC1 and cell death pathways can ultimately guide the development of novel therapies for the treatment of relevant pathological conditions.
Collapse
Affiliation(s)
- Jiajun Zhu
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China,Tsinghua-Peking Center for Life Sciences, Beijing, China,Correspondence to Jiajun Zhu:
| | - Hua Wang
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Xuejun Jiang
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY,Xuejun Jiang:
| |
Collapse
|
20
|
Wolff DW, Bianchi-Smiraglia A, Nikiforov MA. Compartmentalization and regulation of GTP in control of cellular phenotypes. Trends Mol Med 2022; 28:758-769. [PMID: 35718686 PMCID: PMC9420775 DOI: 10.1016/j.molmed.2022.05.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 10/18/2022]
Abstract
Genetic or pharmacological inhibition of enzymes involved in GTP biosynthesis has substantial biological effects, underlining the need to better understand the function of GTP levels in regulation of cellular processes and the significance of targeting GTP biosynthesis enzymes for therapeutic intervention. Our current understanding of spatiotemporal regulation of GTP metabolism and its role in physiological and pathological cellular processes is far from complete. Novel methodologies such as genetically encoded sensors of free GTP offered insights into intracellular distribution and function of GTP molecules. In the current Review, we provide analysis of recent discoveries in the field of GTP metabolism and evaluate the key enzymes as molecular targets.
Collapse
Affiliation(s)
- David W Wolff
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA.
| | - Anna Bianchi-Smiraglia
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| | - Mikhail A Nikiforov
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA; Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
21
|
Liu J, Hong S, Yang J, Zhang X, Wang Y, Wang H, Peng J, Hong L. Targeting purine metabolism in ovarian cancer. J Ovarian Res 2022; 15:93. [PMID: 35964092 PMCID: PMC9375293 DOI: 10.1186/s13048-022-01022-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/17/2022] [Indexed: 11/10/2022] Open
Abstract
Purine, an abundant substrate in organisms, is a critical raw material for cell proliferation and an important factor for immune regulation. The purine de novo pathway and salvage pathway are tightly regulated by multiple enzymes, and dysfunction in these enzymes leads to excessive cell proliferation and immune imbalance that result in tumor progression. Maintaining the homeostasis of purine pools is an effective way to control cell growth and tumor evolution, and exploiting purine metabolism to suppress tumors suggests interesting directions for future research. In this review, we describe the process of purine metabolism and summarize the role and potential therapeutic effects of the major purine-metabolizing enzymes in ovarian cancer, including CD39, CD73, adenosine deaminase, adenylate kinase, hypoxanthine guanine phosphoribosyltransferase, inosine monophosphate dehydrogenase, purine nucleoside phosphorylase, dihydrofolate reductase and 5,10-methylenetetrahydrofolate reductase. Purinergic signaling is also described. We then provide an overview of the application of purine antimetabolites, comprising 6-thioguanine, 6-mercaptopurine, methotrexate, fludarabine and clopidogrel. Finally, we discuss the current challenges and future opportunities for targeting purine metabolism in the treatment-relevant cellular mechanisms of ovarian cancer.
Collapse
Affiliation(s)
- Jingchun Liu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shasha Hong
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiang Yang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoyi Zhang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ying Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Haoyu Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiaxin Peng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Li Hong
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
22
|
Guo C, Tang Y, Yang Z, Li G, Zhang Y. Hallmark-guided subtypes of hepatocellular carcinoma for the identification of immune-related gene classifiers in the prediction of prognosis, treatment efficacy, and drug candidates. Front Immunol 2022; 13:958161. [PMID: 36032071 PMCID: PMC9399518 DOI: 10.3389/fimmu.2022.958161] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC), accounting for ~90% of all primary liver cancer, is a prevalent malignancy worldwide. The intratumor heterogeneity of its causative etiology, histology, molecular landscape, and immune phenotype makes it difficult to precisely recognize individuals with high mortality risk or tumor-intrinsic treatment resistance, especially immunotherapy. Herein, we comprehensively evaluated the activities of cancer hallmark gene sets and their correlations with the prognosis of HCC patients using gene set variation analysis (GSVA) and identified two HCC subtypes with distinct prognostic outcomes. Based on these subtypes, seven immune-related genes (TMPRSS6, SPP1, S100A9, EPO, BIRC5, PLXNA1, and CDK4) were used to construct a novel prognostic gene signature [hallmark-guided subtypes-based immunologic signature (HGSIS)] via multiple statistical approaches. The HGSIS-integrated nomogram suggested an enhanced predictive performance. Interestingly, oncogenic hallmark pathways were significantly enriched in the high-risk group and positively associated with the risk score. Distinct mutational landscapes and immune profiles were observed between different risk groups. Moreover, immunophenoscore (IPS) and tumor immune dysfunction and exclusion (TIDE) analysis showed different sensitivities of HGSIS risk groups for immune therapy efficacy, and the pRRophetic algorithm indicated distinguishable responses for targeted/chemotherapies in different groups. KIF2C was picked out as the key target concerning HGSIS, and the top 10 small molecules were predicted to bind to the active site of KIF2C via molecular docking, which might be further used for candidate drug discovery of HCC. Taken together, our study offers novel insights for clinically significant subtype recognition, and the proposed signature may be a helpful guide for clinicians to improve the treatment regimens.
Collapse
Affiliation(s)
- Chengbin Guo
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yuqin Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhao Yang
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Gen Li
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yongqiang Zhang
- Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
23
|
Dutta N, Deb I, Sarzynska J, Lahiri A. Inosine and its methyl derivatives: Occurrence, biogenesis, and function in RNA. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2022; 169-170:21-52. [PMID: 35065168 DOI: 10.1016/j.pbiomolbio.2022.01.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 12/11/2021] [Accepted: 01/11/2022] [Indexed: 05/21/2023]
Abstract
Inosine is one of the most common post-transcriptional modifications. Since its discovery, it has been noted for its ability to contribute to non-Watson-Crick interactions within RNA. Rapidly accumulating evidence points to the widespread generation of inosine through hydrolytic deamination of adenosine to inosine by different classes of adenosine deaminases. Three naturally occurring methyl derivatives of inosine, i.e., 1-methylinosine, 2'-O-methylinosine and 1,2'-O-dimethylinosine are currently reported in RNA modification databases. These modifications are expected to lead to changes in the structure, folding, dynamics, stability and functions of RNA. The importance of the modifications is indicated by the strong conservation of the modifying enzymes across organisms. The structure, binding and catalytic mechanism of the adenosine deaminases have been well-studied, but the underlying mechanism of the catalytic reaction is not very clear yet. Here we extensively review the existing data on the occurrence, biogenesis and functions of inosine and its methyl derivatives in RNA. We also included the structural and thermodynamic aspects of these modifications in our review to provide a detailed and integrated discussion on the consequences of A-to-I editing in RNA and the contribution of different structural and thermodynamic studies in understanding its role in RNA. We also highlight the importance of further studies for a better understanding of the mechanisms of the different classes of deamination reactions. Further investigation of the structural and thermodynamic consequences and functions of these modifications in RNA should provide more useful information about their role in different diseases.
Collapse
Affiliation(s)
- Nivedita Dutta
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92, Acharya Prafulla Chandra Road, Kolkata, 700009, West Bengal, India
| | - Indrajit Deb
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92, Acharya Prafulla Chandra Road, Kolkata, 700009, West Bengal, India
| | - Joanna Sarzynska
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704, Poznan, Poland
| | - Ansuman Lahiri
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92, Acharya Prafulla Chandra Road, Kolkata, 700009, West Bengal, India.
| |
Collapse
|
24
|
Shlomovitz O, Ben-Zeev B, Pleniceanu O, Greenberger S, Lahav E, Mini S, Tzadok M. An Israeli tuberous sclerosis cohort: the efficacy of different anti-epileptic strategies. Childs Nerv Syst 2021; 37:3827-3833. [PMID: 34491422 DOI: 10.1007/s00381-021-05348-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 08/27/2021] [Indexed: 11/25/2022]
Abstract
AIM We aimed to describe the experience of a large single-center cohort for the clinical, radiological, and genetic characteristics, as well as to determine the efficacy of different anti-epileptic strategies in children and adults with tuberous sclerosis complex (TSC). METHODS We carried out a historical cohort study on 91 TSC patients treated in a single center between 2008 and 2018. RESULTS Our cohort comprised 46 males and 45 females, with a median age of 15.6 years at the last follow-up. Mean follow-up time was 2.5 ± 0.75-5.5 years (range 0-9.5 years). Of those tested, a disease-causing mutation was identified in 90% of patients, 53% in TSC2, and 37% in TSC1. Epilepsy prevalence was similar among TSC1 and TSC2 mutated patients. The most common radiological finding were cortical tubers in 95% of patients, while subependymal giant cell astrocytoma (SEGA) were detected in 36% of patients. Notably, infantile spasms (IS) were diagnosed in 29%, with SEGA representing the only finding significantly different in prevalence between those with and without IS (62% vs. 28%, respectively, p = 0.009). Lastly, we did not find any difference in efficacy between three anti-epileptic treatments: Vagus nerve stimulation (VNS), CBD-based products, and the ketogenic diet, all showing approximately 30%-40% response rates. SIGNIFICANCE Altogether, we provide a comprehensive description of our experience in treating TSC, which could serve to expand current knowledge of the disease and its treatments.
Collapse
Affiliation(s)
- Omer Shlomovitz
- Department of Pediatrics B, Sheba Medical Center, Tel-Hashomer and Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| | - Bruria Ben-Zeev
- Pediatric Neurology Units, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Ramat Gan, Israel
| | - Oren Pleniceanu
- Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shoshana Greenberger
- Dermatology Department, Pediatric Dermatology Service, Sheba Medical Center, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Einat Lahav
- Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Ramat Gan, Israel
| | - Sharon Mini
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- The Institute of Nephrology and Hypertension, Sheba medical center, Ramat Gan, Israel
| | - Michal Tzadok
- Pediatric Neurology Units, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Ramat Gan, Israel
| |
Collapse
|
25
|
Abstract
Tumour initiation and progression requires the metabolic reprogramming of cancer cells. Cancer cells autonomously alter their flux through various metabolic pathways in order to meet the increased bioenergetic and biosynthetic demand as well as mitigate oxidative stress required for cancer cell proliferation and survival. Cancer driver mutations coupled with environmental nutrient availability control flux through these metabolic pathways. Metabolites, when aberrantly accumulated, can also promote tumorigenesis. The development and application of new technologies over the last few decades has not only revealed the heterogeneity and plasticity of tumours but also allowed us to uncover new metabolic pathways involved in supporting tumour growth. The tumour microenvironment (TME), which can be depleted of certain nutrients, forces cancer cells to adapt by inducing nutrient scavenging mechanisms to sustain cancer cell proliferation. There is growing appreciation that the metabolism of cell types other than cancer cells within the TME, including endothelial cells, fibroblasts and immune cells, can modulate tumour progression. Because metastases are a major cause of death of patients with cancer, efforts are underway to understand how metabolism is harnessed by metastatic cells. Additionally, there is a new interest in exploiting cancer genetic analysis for patient stratification and/or dietary interventions in combination with therapies that target metabolism. In this Perspective, we highlight these main themes that are currently under investigation in the context of in vivo tumour metabolism, specifically emphasizing questions that remain unanswered.
Collapse
Affiliation(s)
| | - Navdeep S Chandel
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
26
|
Huang F, Huffman KE, Wang Z, Wang X, Li K, Cai F, Yang C, Cai L, Shih TS, Zacharias LG, Chung A, Yang Q, Chalishazar MD, Ireland AS, Stewart CA, Cargill K, Girard L, Liu Y, Ni M, Xu J, Wu X, Zhu H, Drapkin B, Byers LA, Oliver TG, Gazdar AF, Minna JD, DeBerardinis RJ. Guanosine triphosphate links MYC-dependent metabolic and ribosome programs in small-cell lung cancer. J Clin Invest 2021; 131:139929. [PMID: 33079728 PMCID: PMC7773395 DOI: 10.1172/jci139929] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 10/15/2020] [Indexed: 12/21/2022] Open
Abstract
MYC stimulates both metabolism and protein synthesis, but how cells coordinate these complementary programs is unknown. Previous work reported that, in a subset of small-cell lung cancer (SCLC) cell lines, MYC activates guanosine triphosphate (GTP) synthesis and results in sensitivity to inhibitors of the GTP synthesis enzyme inosine monophosphate dehydrogenase (IMPDH). Here, we demonstrated that primary MYChi human SCLC tumors also contained abundant guanosine nucleotides. We also found that elevated MYC in SCLCs with acquired chemoresistance rendered these otherwise recalcitrant tumors dependent on IMPDH. Unexpectedly, our data indicated that IMPDH linked the metabolic and protein synthesis outputs of oncogenic MYC. Coexpression analysis placed IMPDH within the MYC-driven ribosome program, and GTP depletion prevented RNA polymerase I (Pol I) from localizing to ribosomal DNA. Furthermore, the GTPases GPN1 and GPN3 were upregulated by MYC and directed Pol I to ribosomal DNA. Constitutively GTP-bound GPN1/3 mutants mitigated the effect of GTP depletion on Pol I, protecting chemoresistant SCLC cells from IMPDH inhibition. GTP therefore functioned as a metabolic gate tethering MYC-dependent ribosome biogenesis to nucleotide sufficiency through GPN1 and GPN3. IMPDH dependence is a targetable vulnerability in chemoresistant MYChi SCLC.
Collapse
Affiliation(s)
- Fang Huang
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Children’s Medical Center Research Institute
| | - Kenneth E. Huffman
- Hamon Center for Therapeutic Oncology Research, Departments of Internal Medicine and Pharmacology, and Simmons Comprehensive Cancer Center
| | - Zixi Wang
- Children’s Medical Center Research Institute
| | - Xun Wang
- Children’s Medical Center Research Institute
| | - Kailong Li
- Children’s Medical Center Research Institute
| | - Feng Cai
- Children’s Medical Center Research Institute
| | | | - Ling Cai
- Children’s Medical Center Research Institute
- Department of Population and Data Sciences, and
| | | | | | | | - Qian Yang
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Milind D. Chalishazar
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - Abbie S. Ireland
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - C. Allison Stewart
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kasey Cargill
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Luc Girard
- Hamon Center for Therapeutic Oncology Research, Departments of Internal Medicine and Pharmacology, and Simmons Comprehensive Cancer Center
| | - Yi Liu
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Min Ni
- Children’s Medical Center Research Institute
| | - Jian Xu
- Children’s Medical Center Research Institute
| | - Xudong Wu
- Department of Cell Biology, Tianjin Medical University, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, Tianjin, China
| | - Hao Zhu
- Children’s Medical Center Research Institute
| | - Benjamin Drapkin
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Lauren A. Byers
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Trudy G. Oliver
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - Adi F. Gazdar
- Hamon Center for Therapeutic Oncology Research, Departments of Internal Medicine and Pharmacology, and Simmons Comprehensive Cancer Center
| | - John D. Minna
- Hamon Center for Therapeutic Oncology Research, Departments of Internal Medicine and Pharmacology, and Simmons Comprehensive Cancer Center
| | - Ralph J. DeBerardinis
- Children’s Medical Center Research Institute
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
27
|
Benjanuwattra J, Chaiyawat P, Pruksakorn D, Koonrungsesomboon N. Therapeutic potential and molecular mechanisms of mycophenolic acid as an anticancer agent. Eur J Pharmacol 2020; 887:173580. [PMID: 32949604 DOI: 10.1016/j.ejphar.2020.173580] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 12/14/2022]
Abstract
Mycophenolic acid (MPA) is the active metabolite of mycophenolate mofetil (MMF), an immunosuppressive drug approved for the prophylaxis of allograft rejection in transplant recipients. Recent advances in the role of the type II isoform of inosine-5'-monophosphate dehydrogenase (IMPDH2) in the tumorigenesis of various types of cancer have called for a second look of MPA, the first IMPDH2 inhibitor discovered a hundred years ago, to be repurposed as an anticancer agent. Over a half century, a number of in vitro and in vivo experiments have consistently shown anticancer activity of MPA against several cell lines obtained from different malignancies and murine models. However, a few clinical trials have been conducted to investigate its anticancer activity in humans, and most of which have shown unsatisfactory results. Understanding of available evidence and underlying mechanism of action is a key step to be done so as to facilitate further investigations of MPA to reach its full therapeutic potential as an anticancer agent. This article provides a comprehensive review of non-clinical and clinical evidence available to date, with the emphasis on the molecular mechanism of action in which MPA exerts its anticancer activities: induction of apoptosis, induction of cell cycle arrest, and alteration of tumor microenvironment. Future perspective for further development of MPA to be an anticancer agent is extensively discussed, with the aim of translating the anticancer property of MPA from bench to bedside.
Collapse
Affiliation(s)
| | - Parunya Chaiyawat
- Musculoskeletal Science and Translational Research Center (MSTR), Faculty of Medicine, Chiang Mai University, Thailand; Omics Center for Health Sciences (OCHS), Faculty of Medicine, Chiang Mai University, Thailand
| | - Dumnoensun Pruksakorn
- Musculoskeletal Science and Translational Research Center (MSTR), Faculty of Medicine, Chiang Mai University, Thailand; Omics Center for Health Sciences (OCHS), Faculty of Medicine, Chiang Mai University, Thailand; Biomedical Engineering Institute, Chiang Mai University, Thailand
| | - Nut Koonrungsesomboon
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Thailand; Musculoskeletal Science and Translational Research Center (MSTR), Faculty of Medicine, Chiang Mai University, Thailand.
| |
Collapse
|